Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Front Cell Infect Microbiol ; 12: 900848, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928205

RESUMO

Both, antibiotic persistence and antibiotic resistance characterize phenotypes of survival in which a bacterial cell becomes insensitive to one (or even) more antibiotic(s). However, the molecular basis for these two antibiotic-tolerant phenotypes is fundamentally different. Whereas antibiotic resistance is genetically determined and hence represents a rather stable phenotype, antibiotic persistence marks a transient physiological state triggered by various stress-inducing conditions that switches back to the original antibiotic sensitive state once the environmental situation improves. The molecular basics of antibiotic resistance are in principle well understood. This is not the case for antibiotic persistence. Under all culture conditions, there is a stochastically formed, subpopulation of persister cells in bacterial populations, the size of which depends on the culture conditions. The proportion of persisters in a bacterial population increases under different stress conditions, including treatment with bactericidal antibiotics (BCAs). Various models have been proposed to explain the formation of persistence in bacteria. We recently hypothesized that all physiological culture conditions leading to persistence converge in the inability of the bacteria to re-initiate a new round of DNA replication caused by an insufficient level of the initiator complex ATP-DnaA and hence by the lack of formation of a functional orisome. Here, we extend this hypothesis by proposing that in this persistence state the bacteria become more susceptible to mutation-based antibiotic resistance provided they are equipped with error-prone DNA repair functions. This is - in our opinion - in particular the case when such bacterial populations are exposed to BCAs.


Assuntos
Antibacterianos , Bactérias , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias/genética , Farmacorresistência Bacteriana , Resistência Microbiana a Medicamentos
2.
Front Cell Infect Microbiol ; 10: 615450, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33520740

RESUMO

Persistence has evolved as a potent survival strategy to overcome adverse environmental conditions. This capability is common to almost all bacteria, including all human bacterial pathogens and likely connected to chronic infections caused by some of these pathogens. Although the majority of a bacterial cell population will be killed by the particular stressors, like antibiotics, oxygen and nitrogen radicals, nutrient starvation and others, a varying subpopulation (termed persisters) will withstand the stress situation and will be able to revive once the stress is removed. Several factors and pathways have been identified in the past that apparently favor the formation of persistence, such as various toxin/antitoxin modules or stringent response together with the alarmone (p)ppGpp. However, persistence can occur stochastically in few cells even of stress-free bacterial populations. Growth of these cells could then be induced by the stress conditions. In this review, we focus on the persister formation of human intracellular bacterial pathogens, some of which belong to the most successful persister producers but lack some or even all of the assumed persistence-triggering factors and pathways. We propose a mechanism for the persister formation of these bacterial pathogens which is based on their specific intracellular bipartite metabolism. We postulate that this mode of metabolism ultimately leads, under certain starvation conditions, to the stalling of DNA replication initiation which may be causative for the persister state.


Assuntos
Antitoxinas , Escherichia coli , Antibacterianos/farmacologia , Bactérias , Humanos
3.
Artigo em Inglês | MEDLINE | ID: mdl-30886834

RESUMO

Viruses and intracellular bacterial pathogens (IBPs) have in common the need of suitable host cells for efficient replication and proliferation during infection. In human infections, the cell types which both groups of pathogens are using as hosts are indeed quite similar and include phagocytic immune cells, especially monocytes/macrophages (MOs/MPs) and dendritic cells (DCs), as well as nonprofessional phagocytes, like epithelial cells, fibroblasts and endothelial cells. These terminally differentiated cells are normally in a metabolically quiescent state when they are encountered by these pathogens during infection. This metabolic state of the host cells does not meet the extensive need for nutrients required for efficient intracellular replication of viruses and especially IBPs which, in contrast to the viral pathogens, have to perform their own specific intracellular metabolism to survive and efficiently replicate in their host cell niches. For this goal, viruses and IBPs have to reprogram the host cell metabolism in a pathogen-specific manner to increase the supply of nutrients, energy, and metabolites which have to be provided to the pathogen to allow its replication. In viral infections, this appears to be often achieved by the interaction of specific viral factors with central metabolic regulators, including oncogenes and tumor suppressors, or by the introduction of virus-specific oncogenes. Less is so far known on the mechanisms leading to metabolic reprogramming of the host cell by IBPs. However, the still scant data suggest that similar mechanisms may also determine the reprogramming of the host cell metabolism in IBP infections. In this review, we summarize and compare the present knowledge on this important, yet still poorly understood aspect of pathogenesis of human viral and especially IBP infections.


Assuntos
Bactérias/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Metabolismo , Vírus/crescimento & desenvolvimento , Animais , Regulação da Expressão Gênica , Humanos
4.
Front Immunol ; 9: 2599, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30581430

RESUMO

Objective: Postoperative ileus (POI) is an inflammation-mediated complication of abdominal surgery, characterized by intestinal dysmotility and leukocyte infiltration into the muscularis externa (ME). Previous studies indicated that interleukin (IL)-10 is crucial for the resolution of a variety of inflammation-driven diseases. Herein, we investigated how IL-10 affects the postoperative ME inflammation and found an unforeseen role of IL-10 in POI. Design: POI was induced by a standardized intestinal manipulation (IM) in C57BL/6 and multiple transgenic mouse strain including C-C motif chemokine receptor 2-/-, IL-10-/-, and LysMcre/IL-10fl/fl mice. Leukocyte infiltration, gene and protein expression of cytokines, chemokines, and macrophage differentiation markers as well as intestinal motility were analyzed. IL-10 serum levels in surgical patients were determined by ELISA. Results: IL-10 serum levels were increased in patient after abdominal surgery. In mice, a complete or leucocyte-restricted IL-10 deficiency ameliorated POI and reduced the postoperative ME neutrophil infiltration. Infiltrating monocytes were identified as main IL-10 producers and undergo IL-10-dependent M2 polarization. Interestingly, M2 polarization is not crucial to POI development as abrogation of monocyte infiltration did not prevent POI due to a compensation of the IL-10 loss by resident macrophages and neutrophils. Organ culture studies demonstrated that IL-10 deficiency impeded neutrophil migration toward the surgically traumatized ME. This mechanism is mediated by reduction of neutrophil attracting chemokines. Conclusion: Monocyte-derived macrophages are the major IL-10 source during POI. An IL-10 deficiency decreases the postoperative expression of neutrophil-recruiting chemokines, consequently reduces the neutrophil extravasation into the postsurgical bowel wall, and finally protects mice from POI.


Assuntos
Íleus/imunologia , Interleucina-10/imunologia , Intestinos/imunologia , Leucócitos/imunologia , Complicações Pós-Operatórias/imunologia , Animais , Modelos Animais de Doenças , Motilidade Gastrointestinal/imunologia , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia , Monócitos/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Período Pós-Operatório
5.
Artigo em Inglês | MEDLINE | ID: mdl-28752080

RESUMO

Intracellular bacterial pathogens (IBPs) invade and replicate in different cell types including immune cells, in particular of the innate immune system (IIS) during infection in the acute phase. However, immune cells primarily function as essential players in the highly effective and integrated host defense systems comprising the IIS and the adaptive immune system (AIS), which cooperatively protect the host against invading microbes including IBPs. As countermeasures, the bacterial pathogens (and in particular the IBPs) have developed strategies to evade or reprogram the IIS at various steps. The intracellular replication capacity and the anti-immune defense responses of the IBP's as well as the specific antimicrobial responses of the immune cells of the innate and the AIS depend on specific metabolic programs of the IBPs and their host cells. The metabolic programs of the immune cells supporting or counteracting replication of the IBPs appear to be mutually exclusive. Indeed, recent studies show that upon interaction of naïve, metabolically quiescent immune cells with IBPs, different metabolic activation processes occur which may result in the provision of a survival and replication niche for the pathogen or its eradication. It is therefore likely that within a possible host cell population subsets exist that are metabolically programmed for pro- or anti-microbial conditions. These metabolic programs may be triggered by the interactions between different bacterial agonistic components and host cell receptors. In this review, we summarize the current status in the field and discuss metabolic adaptation processes within immune cells of the IIS and the IBPs that support or restrict the intracellular replication of the pathogens.


Assuntos
Bactérias/metabolismo , Infecções Bacterianas/metabolismo , Células Sanguíneas/metabolismo , Animais , Bactérias/genética , Bactérias/imunologia , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Células Sanguíneas/imunologia , Células Sanguíneas/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata
6.
Sci Rep ; 6: 29275, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27387855

RESUMO

In Enterobacteriaceae, the RNA chaperone Hfq mediates the interaction of small RNAs with target mRNAs, thereby modulating transcript stability and translation. This post-transcriptional control helps bacteria adapt quickly to changing environmental conditions. Our previous mutational analysis showed that Hfq is involved in metabolism and stress survival in the enteropathogen Yersinia enterocolitica. In this study we demonstrate that Hfq is essential for virulence in mice and influences production of surface pathogenicity factors, in particular lipopolysaccharide and adhesins mediating interaction with host tissue. Hfq inhibited the production of Ail, the Ail-like protein OmpX and the MyfA pilin post-transcriptionally. In contrast Hfq promoted production of two major autotransporter adhesins YadA and InvA. While protein secretion in vitro was not affected, hfq mutants exhibited decreased protein translocation by the type III secretion system into host cells, consistent with decreased production of YadA and InvA. The influence of Hfq on YadA resulted from a complex interplay of transcriptional, post-transcriptional and likely post-translational effects. Hfq regulated invA by modulating the expression of the transcriptional regulators rovA, phoP and ompR. Therefore, Hfq is a global coordinator of surface virulence determinants in Y. enterocolitica suggesting that it constitutes an attractive target for developing new antimicrobial strategies.


Assuntos
Adesinas Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica , Fator Proteico 1 do Hospedeiro/metabolismo , Fatores de Virulência/metabolismo , Yersinia enterocolitica/patogenicidade , Animais , Modelos Animais de Doenças , Camundongos , Virulência , Yersiniose/microbiologia , Yersiniose/patologia , Yersinia enterocolitica/metabolismo
7.
Environ Microbiol ; 18(3): 997-1021, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26627632

RESUMO

Enteropathogenic Yersinia enterocolitica is able to grow within or outside the mammalian host. Previous transcriptomic studies have indicated that the regulator OmpR plays a role in the expression of hundreds of genes in enterobacteria. Here, we have examined the impact of OmpR on the production of Y. enterocolitica membrane proteins upon changes in temperature, osmolarity and pH. Proteomic analysis indicated that the loss of OmpR affects the production of 120 proteins, a third of which are involved in uptake/transport, including several that participate in iron or heme acquisition. A set of proteins associated with virulence was also affected. The influence of OmpR on the abundance of adhesin YadA and heme receptor HemR was examined in more detail. OmpR was found to repress YadA production and bind to the yadA promoter, suggesting a direct regulatory effect. In contrast, the repression of hemR expression by OmpR appears to be indirect. These findings provide new insights into the role of OmpR in remodelling the cell surface and the adaptation of Y. enterocolitica to different environmental niches, including the host.


Assuntos
Adesinas Bacterianas/biossíntese , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Receptores de Superfície Celular/biossíntese , Transativadores/genética , Yersinia enterocolitica/genética , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/genética , Dados de Sequência Molecular , Concentração Osmolar , Regiões Promotoras Genéticas , Proteoma/metabolismo , Proteômica , Receptores de Superfície Celular/genética , Virulência
8.
Appl Environ Microbiol ; 81(20): 7041-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26231647

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) is the causative agent of bloody diarrhea and extraintestinal sequelae in humans, most importantly hemolytic-uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). Besides the bacteriophage-encoded Shiga toxin gene (stx), EHEC harbors the locus of enterocyte effacement (LEE), which confers the ability to cause attaching and effacing lesions. Currently, the vast majority of EHEC infections are caused by strains belonging to five O serogroups (the "big five"), which, in addition to O157, the most important, comprise O26, O103, O111, and O145. We hypothesize that these four non-O157 EHEC serotypes differ in their phylogenies. To test this hypothesis, we used multilocus sequence typing (MLST) to analyze a large collection of 250 isolates of these four O serogroups, which were isolated from diseased as well as healthy humans and cattle between 1952 and 2009. The majority of the EHEC isolates of O serogroups O26 and O111 clustered into one sequence type complex, STC29. Isolates of O103 clustered mainly in STC20, and most isolates of O145 were found within STC32. In addition to these EHEC strains, STC29 also included stx-negative E. coli strains, termed atypical enteropathogenic E. coli (aEPEC), yet another intestinal pathogenic E. coli group. The finding that aEPEC and EHEC isolates of non-O157 O serogroups share the same phylogeny suggests an ongoing microevolutionary scenario in which the phage-encoded Shiga toxin gene stx is transferred between aEPEC and EHEC. As a consequence, aEPEC strains of STC29 can be regarded as post- or pre-EHEC isolates. Therefore, STC29 incorporates phylogenetic information useful for unraveling the evolution of EHEC.


Assuntos
Escherichia coli Êntero-Hemorrágica/classificação , Escherichia coli Êntero-Hemorrágica/genética , Evolução Molecular , Genótipo , Filogenia , Sorogrupo , Animais , Bovinos , Análise por Conglomerados , Colífagos/genética , Escherichia coli Êntero-Hemorrágica/isolamento & purificação , Escherichia coli Enteropatogênica/classificação , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/isolamento & purificação , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/veterinária , Voluntários Saudáveis , Humanos , Tipagem de Sequências Multilocus , Toxinas Shiga/genética
9.
Genome Announc ; 3(4)2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26139712

RESUMO

Pseudomonas aeruginosa is an opportunistic pathogen that typically infects patients with a compromised immune defense. Here, we present the improved 6.5-Mb draft genome of strain WS136, an ExoS-positive and ExoU-negative highly cytotoxic chronic wound isolate recovered from pyoderma gangrenosum of a patient who received bone marrow transplantation.

10.
Microbiol Spectr ; 3(3)2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26185075

RESUMO

Several bacterial pathogens that cause severe infections in warm-blooded animals, including humans, have the potential to actively invade host cells and to efficiently replicate either in the cytosol or in specialized vacuoles of the mammalian cells. The interaction between these intracellular bacterial pathogens and the host cells always leads to multiple physiological changes in both interacting partners, including complex metabolic adaptation reactions aimed to promote proliferation of the pathogen within different compartments of the host cells. In this chapter, we discuss the necessary nutrients and metabolic pathways used by some selected cytosolic and vacuolar intracellular pathogens and--when available--the links between the intracellular bacterial metabolism and the expression of the virulence genes required for the intracellular bacterial replication cycle. Furthermore, we address the growing evidence that pathogen-specific factors may also trigger metabolic responses of the infected mammalian cells affecting the carbon and nitrogen metabolism as well as defense reactions. We also point out that many studies on the metabolic host cell responses induced by the pathogens have to be scrutinized due to the use of established cell lines as model host cells, as these cells are (in the majority) cancer cells that exhibit a dysregulated primary carbon metabolism. As the exact knowledge of the metabolic host cell responses may also provide new concepts for antibacterial therapies, there is undoubtedly an urgent need for host cell models that more closely reflect the in vivo infection conditions.


Assuntos
Adaptação Fisiológica/fisiologia , Bactérias/metabolismo , Bactérias/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Espaço Intracelular/microbiologia , Animais , Bactérias/crescimento & desenvolvimento , Carbono/metabolismo , Citosol/microbiologia , Humanos , Nitrogênio/metabolismo , Vacúolos/microbiologia , Fatores de Virulência/metabolismo
11.
Genome Announc ; 2(6)2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25523779

RESUMO

Pseudomonas aeruginosa is a frequent human pathogen that increasingly causes chronic infections of nonhealing wounds. Here we present the 6.8 Mb draft genome of strain WS394, a multidrug-resistant chronic ulcer isolate that exhibited outstanding high cell cytotoxicity despite defective secretion of exotoxin U, suggesting a habitat-dependent adaptation process.

12.
Int J Med Microbiol ; 304(7): 824-34, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25172222

RESUMO

Enteropathogenic Yersinia enterocolitica bioserotype 4/O:3 are the most frequent cause of human yersiniosis worldwide with symptoms ranging from mild diarrhea to severe complications of mesenteric lymphadenitis, liver abscesses and postinfectious extraintestinal sequelae. The main reservoir host of 4/O:3 strains are pigs, which represent a substantial disease-causing potential for humans, as they are usually asymptomatic carriers. Y. enterocolitica O:3 initiates infections by tight attachment to the intestinal mucosa. Colonization of the digestive tract is frequently followed by invasion of the intestinal layer primarily at the follicle-associated epithelium, allowing the bacteria to propagate in the lamina propria and disseminate into deeper tissues. Molecular characterization of Y. enterocolitica O:3 isolates led to the identification of (i) alternative virulence and fitness factors and (ii) small genetic variations which cause profound changes in their virulence gene expression pattern (e.g. constitutive expression of the primary invasion factor InvA). These changes provoke a major difference in the virulence properties, i.e. reduced colonization of intestinal tissues in mice, but improved long-term colonization in the pig intestine. Y. enterocolitica O:3 strains cause also a considerably lower level of proinflammatory cytokine IL-8 and higher levels of the anti-inflammatory cytokine IL-10 in porcine primary macrophages, as compared to murine macrophages, which could contribute to limiting inflammation, immunopathology and severity of the infection in pigs.


Assuntos
Reservatórios de Doenças , Suínos/microbiologia , Yersiniose/veterinária , Yersinia enterocolitica/fisiologia , Animais , Trato Gastrointestinal/microbiologia , Interações Hospedeiro-Patógeno , Interleucina-10/metabolismo , Interleucina-8/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Virulência , Fatores de Virulência/genética , Yersiniose/imunologia , Yersiniose/microbiologia , Yersiniose/patologia , Yersinia enterocolitica/genética
13.
Int J Med Microbiol ; 304(8): 1050-61, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25130702

RESUMO

Pseudomonas aeruginosa is the leading pathogen of chronic cystic fibrosis (CF) lung infection. Life-long persistence in the inflamed and ever fluctuating CF lungs results in the selection of a variety of changes in P. aeruginosa physiology. Accumulating evidence suggests that especially metabolic changes support the survival and growth of P. aeruginosa within the hypoxic and nutritious CF mucus. To investigate if metabolic adaptations we described for hypermutable P. aeruginosa from late CF lung disease (Hoboth et al., 2009. J. Infect. Dis., pp. 118-130) may represent specific changes in response to the selective conditions within the oxygen-restricted CF mucus, we determined the expression of a set of genes during aerobic and hypoxic growth in LB and the artificial sputum medium ASM. We further focused on the regulation of the two isocitrate dehydrogenases Icd and Idh. Interestingly, both isoenzymes may replace each other under aerobic and hypoxic conditions. The NADPH- and RpoS-dependent Icd seems to be the leading isoenzyme under prolonged oxygen limitation and stationary growth phase. LacZ reporter analysis revealed that oxygen-restriction increased the expression levels of azu, cbb3-1, cbb3-2, ccpR, icd, idh and oprF gene, whereas himD and nuoA are increasingly expressed only during hypoxic growth in ASM. Overexpression of the anaerobic regulator Anr improved the expression of azu, ccpR, cbb3-2 and icd. In summary, expression of azu, cbb3-1, cbb3-2, ccpR, icd, idh, oprF, himD, and nuoA appeared to be beneficial for the growth of P. aeruginosa under hypoxic conditions indicating these genes may represent marker genes for the metabolic adaptation to the CF lung environment.


Assuntos
Adaptação Biológica , Fibrose Cística/complicações , Marcadores Genéticos , Hipóxia/complicações , Redes e Vias Metabólicas/genética , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Fibrose Cística/patologia , Expressão Gênica , Humanos , Isocitrato Desidrogenase/genética , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/isolamento & purificação , Pseudomonas aeruginosa/metabolismo , Infecções Respiratórias/microbiologia
14.
Int J Med Microbiol ; 304(3-4): 444-51, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24636859

RESUMO

The Yersinia outer protein M (YopM) is a type 3 secretion system (T3SS)-dependent effector protein of Yersinia enterocolitica, Yersinia pseudotuberculosis and Yersinia pestis. Although YopM is indispensable for full virulence, its molecular functions still remain largely elusive. Recently, we could identify the recombinant YopM (rYopM) protein derived from the Y. enterocolitica strain 8081 (JB580) as a cell-penetrating protein, which down-regulates the expression of various pro-inflammatory cytokines including TNFα. In this study, we have generated rabbit monoclonal anti-YopM antibodies (RabMabs). RabMabs were characterized by SDS-PAGE and Western blotting using various truncated versions of rYopM to identify epitope-containing domains. RabMabs recognizing either the N- or C-terminus of YopM were characterized further and validated using a collection of 61 pathogenic and non-pathogenic Yersinia strains as well as exemplary strains of major intestinal bacterial pathogens such as Salmonella enterica ssp. enterica, Shigella flexneri and intestinal pathogenic Escherichia coli. RabMab 41.3 directed at the N-terminus of YopM of Y. enterocolitica strain 8081 recognized all YopM-expressing pathogenic Yersinia strains analyzed in this study but failed to recognize non-pathogenic isolates. Thus, RabMab 41.3 might be applicable for the detection of pathogenic Yersinia strains.


Assuntos
Anticorpos Monoclonais , Proteínas da Membrana Bacteriana Externa/imunologia , Yersinia enterocolitica/isolamento & purificação , Yersinia pestis/isolamento & purificação , Yersinia pseudotuberculosis/isolamento & purificação , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Feminino , Coelhos , Sensibilidade e Especificidade , Yersinia enterocolitica/imunologia , Yersinia pestis/imunologia , Yersinia pseudotuberculosis/imunologia
15.
PLoS One ; 9(1): e86113, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24454955

RESUMO

To adapt to changes in environmental conditions, bacteria regulate their gene expression at the transcriptional but also at the post-transcriptional level, e.g. by small RNAs (sRNAs) which modulate mRNA stability and translation. The conserved RNA chaperone Hfq mediates the interaction of many sRNAs with their target mRNAs, thereby playing a global role in fine-tuning protein production. In this study, we investigated the significance of Hfq for the enteropathogen Yersina enterocolitica serotype O:8. Hfq facilitated optimal growth in complex and minimal media. Our comparative protein analysis of parental and hfq-negative strains suggested that Hfq promotes lipid metabolism and transport, cell redox homeostasis, mRNA translation and ATP synthesis, and negatively affects carbon and nitrogen metabolism, transport of siderophore and peptides and tRNA synthesis. Accordingly, biochemical tests indicated that Hfq represses ornithine decarboxylase activity, indole production and utilization of glucose, mannitol, inositol and 1,2-propanediol. Moreover, Hfq repressed production of the siderophore yersiniabactin and its outer membrane receptor FyuA. In contrast, hfq mutants exhibited reduced urease production. Finally, strains lacking hfq were more susceptible to acidic pH and oxidative stress. Unlike previous reports in other Gram-negative bacteria, Hfq was dispensable for type III secretion encoded by the virulence plasmid. Using a chromosomally encoded FLAG-tagged Hfq, we observed increased production of Hfq-FLAG in late exponential and stationary phases. Overall, Hfq has a profound effect on metabolism, resistance to stress and modulates the production of two virulence factors in Y. enterocolitica, namely urease and yersiniabactin.


Assuntos
Fator Proteico 1 do Hospedeiro/fisiologia , Fatores de Virulência/biossíntese , Yersinia enterocolitica/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/biossíntese , Sistemas de Secreção Bacterianos , Bacteriocinas/farmacologia , Metabolismo dos Carboidratos , Testes de Sensibilidade a Antimicrobianos por Disco-Difusão , Técnicas de Inativação de Genes , Indóis/metabolismo , Viabilidade Microbiana , Ornitina Descarboxilase/metabolismo , Estresse Oxidativo , Fenóis/metabolismo , Proteoma/metabolismo , Receptores de Superfície Celular/biossíntese , Tiazóis/metabolismo , Urease/biossíntese , Yersinia enterocolitica/genética , Yersinia enterocolitica/crescimento & desenvolvimento
16.
Int J Med Microbiol ; 304(3-4): 275-83, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24246413

RESUMO

Yersinia enterocolitica is a food-borne, gastro-intestinal pathogen with world-wide distribution. Only 11 serotypes have been isolated from patients, with O:3, O:9, O:8 and O:5,27 being the serotypes most commonly associated with human yersiniosis. Serotype is an important characteristic of Y. enterocolitica strains, allowing differentiation for epidemiology, diagnosis and phylogeny studies. Conventional serotyping, performed by slide agglutination, is a tedious and laborious procedure whose interpretation tends to be subjective, leading to poor reproducibility. Here we present a PCR-based typing scheme for molecular identification and patho-serotyping of Y. enterocolitica. Genome-wide comparison of Y. enterocolitica sequences allowed analysis of the O-antigen gene clusters of different serotypes, uncovering their formerly unknown genomic locations, and selection of targets for serotype-specific amplification. Two multiplex PCRs and one additional PCR were designed and tested on various reference strains and isolates from different origins. Our genotypic assay proved to be highly specific for identification of Y. enterocolitica species, discrimination between virulent and non-virulent strains, distinguishing the main human-related serotypes, and typing of conventionally untypeable strains. This genotyping scheme could be applied in microbiology laboratories as an alternative or complementary method to the traditional phenotypic assays, providing data for epidemiological studies.


Assuntos
Genoma Bacteriano , Técnicas de Genotipagem , Tipagem Molecular/métodos , Yersinia enterocolitica/classificação , Yersinia enterocolitica/genética , Genótipo , Humanos , Família Multigênica , Antígenos O/genética , Reação em Cadeia da Polimerase/métodos , Sorotipagem , Yersiniose/diagnóstico , Yersiniose/microbiologia
17.
FASEB J ; 27(11): 4476-88, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23901070

RESUMO

Antimicrobial peptides are a promising complement to common antibiotics, development of resistance to which is a growing problem. Here we present a de novo-designed peptide, SP1-1 (RKKRLKLLKRLL-NH2), with antimicrobial activity against multiresistant Staphylococcus aureus (minimal inhibitory concentration: 6.25 µM). Elucidation of the mode of action of this peptide revealed a strong interaction with RsbW kinase (Kd: 6.01±2.73 nM), a serine kinase negatively regulating the activity of the transcription factor σB (SigB). SP1-1 binding and functional modulation of RsbW were shown in vitro by a combination of biochemical, molecular, and biophysical methods, which were further genetically evidenced in vivo by analysis of S. aureus ΔsigB deletion mutants. Intracellular localization of the peptide was demonstrated using nanometer-scaled secondary ion mass spectrometry. Moreover, microarray analysis revealed that transcription of numerous genes, involved in cell wall and amino acid metabolism, transport mechanisms, virulence, and pigmentation, is affected. Interestingly, several WalR binding motif containing genes are induced by SP1-1. In sum, the designed peptide SP1-1 seems to have multiple modes of action, including inhibition of a kinase, and therefore might contribute to the development of new antibacterial compounds, giving bacterial kinase inhibition a closer inspection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Transporte/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Staphylococcus aureus/efeitos dos fármacos , Sequência de Aminoácidos , Peptídeos Catiônicos Antimicrobianos/síntese química , Peptídeos Catiônicos Antimicrobianos/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Relação Dose-Resposta a Droga , Dados de Sequência Molecular , Mutação , Ligação Proteica , Inibidores de Proteínas Quinases/síntese química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fator sigma/genética , Fator sigma/metabolismo , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Transcrição Gênica/efeitos dos fármacos , Virulência/genética
18.
Int J Med Microbiol ; 303(8): 433-42, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23810728

RESUMO

Natural Killer (NK) cells serve as an important source of proinflammatory cytokines early during infection. Hypothesizing that Yersinia enterocolitica might interact with and inactivate NK cells, we examined NK cell-Y. enterocolitica interactions in vitro and in vivo. Y. enterocolitica adheres to NK cells in an Invasin dependent manner and inhibits NK cell cytotoxicity and IFN-γ production induced by IL-12+IL-18 or IL-12 alone. YopP, an acetyltransferase known to inhibit MAPK and NFκB signaling, suppresses IL-12 and IL-12+IL-18 mediated IFN-γ production in NK cells by inhibiting phosphorylation of Tyk2 and STAT4 in addition to MAPK. YopP inhibits induction of all genes whose expression is induced by IL-12+IL-18 in NK cells. Y. enterocolitica-mediated adherence to and inactivation of NK cells also occurs after infection in vivo. Thus, we present the first report of a bacterial pathogen inactivating NK cells, and report interaction with Tyk2-STAT4 signaling as a novel function of YopP.


Assuntos
Interações Hospedeiro-Patógeno , Tolerância Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Aderência Bacteriana , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Células Cultivadas , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo , Yersinia enterocolitica/fisiologia
19.
Genome Announc ; 1(4)2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23846271

RESUMO

We report here the complete genome sequences of four European Yersinia enterocolitica mammalian isolates of bioserotype 4/O:3. The genomes have an average size of 4.50 Mb, a G+C content of 47%, and between 4,231 and 4,330 coding sequences (CDSs). No relevant differences were detected by genome comparison between mammalian and human isolates.

20.
Artigo em Inglês | MEDLINE | ID: mdl-23847769

RESUMO

The interaction of bacterial pathogens with mammalian hosts leads to a variety of physiological responses of the interacting partners aimed at an adaptation to the new situation. These responses include multiple metabolic changes in the affected host cells which are most obvious when the pathogen replicates within host cells as in case of intracellular bacterial pathogens. While the pathogen tries to deprive nutrients from the host cell, the host cell in return takes various metabolic countermeasures against the nutrient theft. During this conflicting interaction, the pathogen triggers metabolic host cell responses by means of common cell envelope components and specific virulence-associated factors. These host reactions generally promote replication of the pathogen. There is growing evidence that pathogen-specific factors may interfere in different ways with the complex regulatory network that controls the carbon and nitrogen metabolism of mammalian cells. The host cell defense answers include general metabolic reactions, like the generation of oxygen- and/or nitrogen-reactive species, and more specific measures aimed to prevent access to essential nutrients for the respective pathogen. Accurate results on metabolic host cell responses are often hampered by the use of cancer cell lines that already exhibit various de-regulated reactions in the primary carbon metabolism. Hence, there is an urgent need for cellular models that more closely reflect the in vivo infection conditions. The exact knowledge of the metabolic host cell responses may provide new interesting concepts for antibacterial therapies.


Assuntos
Bactérias/metabolismo , Bactérias/patogenicidade , Interações Hospedeiro-Patógeno , Mamíferos/metabolismo , Mamíferos/microbiologia , Animais , Bactérias/imunologia , Carbono/metabolismo , Regulação da Expressão Gênica , Humanos , Mamíferos/imunologia , Nitrogênio/metabolismo , Espécies Reativas de Nitrogênio/imunologia , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...